Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Transl Res ; 263: 45-52, 2024 01.
Article in English | MEDLINE | ID: mdl-37678755

ABSTRACT

Cyclic nucleotide elevation in intestinal epithelial cells is the key pathology causing intestinal fluid loss in secretory diarrheas such as cholera. Current secretory diarrhea treatment is primarily supportive, and oral rehydration solution is the mainstay of cholera treatment. There is an unmet need for safe, simple and effective diarrhea treatments. By promoting cAMP hydrolysis, extracellular calcium-sensing receptor (CaSR) is a regulator of intestinal fluid transport. We studied the antidiarrheal mechanisms of FDA-approved CaSR activator cinacalcet and tested its efficacy in clinically relevant human cell, mouse and intestinal organoid models of secretory diarrhea. By using selective inhibitors, we found that cAMP agonists-induced secretory short-circuit currents (Isc) in human intestinal T84 cells are mediated by collective actions of apical membrane cystic fibrosis transmembrane conductance regulator (CFTR) and Clc-2 Cl- channels, and basolateral membrane K+ channels. 30 µM cinacalcet pretreatment inhibited all 3 components of forskolin and cholera toxin-induced secretory Isc by ∼75%. In mouse jejunal mucosa, cinacalcet inhibited forskolin-induced secretory Isc by ∼60% in wild type mice, with no antisecretory effect in intestinal epithelia-specific Casr knockout mice (Casr-flox; Vil1-cre). In suckling mouse model of cholera induced by oral cholera toxin, single dose (30 mg/kg) oral cinacalcet treatment reduced intestinal fluid accumulation by ∼55% at 20 hours. Lastly, cinacalcet inhibited forskolin-induced secretory Isc by ∼75% in human colonic and ileal organoids. Our findings suggest that CaSR activator cinacalcet has antidiarrheal efficacy in distinct human cell, organoid and mouse models of secretory diarrhea. Considering its excellent clinical safety profile, cinacalcet can be repurposed as a treatment for cyclic nucleotide-mediated secretory diarrheas including cholera.


Subject(s)
Antidiarrheals , Cholera , Mice , Humans , Animals , Antidiarrheals/metabolism , Antidiarrheals/pharmacology , Antidiarrheals/therapeutic use , Cholera/drug therapy , Cholera/metabolism , Cholera/pathology , Cholera Toxin/metabolism , Cholera Toxin/pharmacology , Cholera Toxin/therapeutic use , Cinacalcet/pharmacology , Cinacalcet/therapeutic use , Cinacalcet/metabolism , Receptors, Calcium-Sensing/metabolism , Receptors, Calcium-Sensing/therapeutic use , Nucleotides, Cyclic/metabolism , Nucleotides, Cyclic/pharmacology , Nucleotides, Cyclic/therapeutic use , Colforsin/metabolism , Colforsin/pharmacology , Colforsin/therapeutic use , Diarrhea/drug therapy , Diarrhea/metabolism , Intestinal Mucosa/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/therapeutic use , Mice, Knockout
2.
FASEB J ; 37(8): e23094, 2023 08.
Article in English | MEDLINE | ID: mdl-37462513

ABSTRACT

Little is known about the effect of the recently developed calcimimetic evocalcet (Evo) on parathyroid calcium-sensing receptor (CaSR) and vitamin D receptor (VDR) expression. We examined the effects of Evo and cinacalcet (Cina) on CaSR and VDR expression in 5/6 nephrectomized Sprague-Dawley rats fed a high-phosphorus diet for 4 weeks to develop secondary hyperparathyroidism (SHPT). These uremic rats were divided into 4 groups-baseline control (Nx4W) and groups with additional treatment with either the Vehicle, Evo, or Cina for 2 weeks; normal rats were used as normal controls (NC). Blood parameters and parathyroid tissue were analyzed. CaSR and VDR expression levels were determined using immunohistochemistry. The degree of kidney injury and hyperphosphatemia was similar in the uremic groups (Nx4W, Vehicle, Cina, and Evo). Serum parathyroid hormone levels were significantly higher in the Nx4W and Vehicle groups than in the NC group. This increase was significantly suppressed in the Cina and Evo groups compared with that in the Vehicle group. Serum calcium levels were significantly and equally lower in the Cina and Evo groups relative to those in the Vehicle group. CaSR expression was significantly lower in the Nx4W and Vehicle groups than in the NC group. This downregulation was of an equally lesser magnitude in the Cina and Evo groups. A similar trend was observed for VDR expression. These results indicate that Evo and Cina treatment can increase parathyroid CaSR and VDR expression in uremic rats with SHPT, which could provide better control of mineral and bone disorder markers.


Subject(s)
Hyperparathyroidism, Secondary , Receptors, Calcitriol , Rats , Animals , Receptors, Calcitriol/metabolism , Receptors, Calcium-Sensing/metabolism , Rats, Sprague-Dawley , Parathyroid Glands/metabolism , Hyperparathyroidism, Secondary/drug therapy , Hyperparathyroidism, Secondary/complications , Hyperparathyroidism, Secondary/metabolism , Parathyroid Hormone/metabolism , Cinacalcet/pharmacology , Cinacalcet/metabolism
3.
Calcif Tissue Int ; 112(4): 452-462, 2023 04.
Article in English | MEDLINE | ID: mdl-36754901

ABSTRACT

Patients with type 2 diabetes mellitus (T2DM) experience a higher risk of fractures despite paradoxically exhibiting normal to high bone mineral density (BMD). This has drawn into question the applicability to T2DM of conventional fracture reduction treatments that aim to retain BMD. In a primary human osteoblast culture system, high glucose levels (25 mM) impaired cell proliferation and matrix mineralization compared to physiological glucose levels (5 mM). Treatment with parathyroid hormone (PTH, 10 nM), a bone anabolic agent, and cinacalcet (CN, 1 µM), a calcimimetic able to target the Ca2+-sensing receptor (CaSR), were tested for their effects on proliferation and differentiation. Strikingly, CN+PTH co-treatment was shown to promote cell growth and matrix mineralization under both physiological and high glucose conditions. CN+PTH reduced apoptosis by 0.9-fold/0.4-fold as measured by Caspase-3 activity assay, increased alkaline phosphatase (ALP) expression by 1.5-fold/twofold, increased the ratio of nuclear factor κ-B ligand (RANKL) to osteoprotegerin (OPG) by 2.1-fold/1.6-fold, and increased CaSR expression by 1.7-fold/4.6-fold (physiological glucose/high glucose). Collectively, these findings indicate a potential for CN+PTH combination therapy as a method to ameliorate the negative impact of chronic high blood glucose on bone remodeling.


Subject(s)
Diabetes Mellitus, Type 2 , Parathyroid Hormone , Humans , Cinacalcet/pharmacology , Cinacalcet/metabolism , Diabetes Mellitus, Type 2/metabolism , Osteoblasts/metabolism , Osteoprotegerin/metabolism , Glucose/metabolism , RANK Ligand/metabolism , Cells, Cultured
4.
Chin J Physiol ; 65(6): 277-281, 2022.
Article in English | MEDLINE | ID: mdl-36588353

ABSTRACT

Ca2+-sensing receptors (CaSRs) are G protein-coupled receptors activated by elevated concentrations of extracellular Ca2+. In our previous works, we showed protein and functional expression of CaSR in mouse cerebral endothelial cell (EC) (bEND.3); the CaSR response (high Ca2+-elicited cytosolic [Ca2+] elevation) was unaffected by suppression of phospholipase C but in part involved Ca2+ influx through transient receptor potential V1 (TRPV1) channels. In this work, we investigated if extracellular acidity affected CaSR-mediated Ca2+ influx triggered by high (3 mM) Ca2+ (CaSR agonist), 3 mM spermine (CaSR agonist), and 10 mM cinacalcet (positive allosteric modulator of CaSR). Extracellular acidosis (pH 6.8 and pH 6.0) strongly suppressed cytosolic [Ca2+] elevation triggered by high Ca2+, spermine, and cinacalcet; acidosis also inhibited Mn2+ influx stimulated by high Ca2+ and cinacalcet. Purinoceptor-triggered Ca2+ response, however, was not suppressed by acidosis. Extracellular acidity also did not affect membrane potential, suggesting suppressed CaSR-mediated Ca2+ influx in acidity did not result from the reduced electrical driving force for Ca2+. Our results suggest Ca2+ influx through a putative CaSR-TRP complex in bEND.3 EC was sensitive to extracellular pH.


Subject(s)
Calcium Signaling , Endothelial Cells , Mice , Animals , Endothelial Cells/metabolism , Cinacalcet/pharmacology , Cinacalcet/metabolism , Spermine/pharmacology , Spermine/metabolism , Membrane Potentials , Calcium/metabolism
5.
JCI Insight ; 6(4)2021 02 22.
Article in English | MEDLINE | ID: mdl-33400691

ABSTRACT

Diarrhea is a major cause of global mortality, and outbreaks of secretory diarrhea such as cholera remain an important problem in the developing world. Current treatment of secretory diarrhea primarily involves supportive measures, such as fluid replacement. The calcium-sensing receptor (CaSR) regulates multiple biological activities in response to changes in extracellular Ca2+. The FDA-approved drug cinacalcet is an allosteric activator of CaSR used for treatment of hyperparathyroidism. Here, we found by short-circuit current measurements in human colonic T84 cells that CaSR activation by cinacalcet reduced forskolin-induced Cl- secretion by greater than 80%. Cinacalcet also reduced Cl- secretion induced by cholera toxin, heat-stable E. coli enterotoxin, and vasoactive intestinal peptide (VIP). The cinacalcet effect primarily involved indirect inhibition of cystic fibrosis transmembrane conductance regulator-mediated (CFTR-mediated) Cl- secretion following activation of CaSR and downstream phospholipase C and phosphodiesterases. In mice, cinacalcet reduced fluid accumulation by more than 60% in intestinal closed loop models of cholera and traveler's diarrhea. The cinacalcet effect involved both inhibition of CFTR-mediated secretion and stimulation of sodium-hydrogen exchanger 3-mediated absorption. These findings support the therapeutic utility of the safe and commonly used drug cinacalcet in CFTR-dependent secretory diarrheas, including cholera, traveler's diarrhea, and VIPoma.


Subject(s)
Cinacalcet/therapeutic use , Cystic Fibrosis Transmembrane Conductance Regulator/adverse effects , Diarrhea/drug therapy , Drug Repositioning/methods , Receptors, Calcium-Sensing/therapeutic use , Animals , Bacterial Toxins , Cell Line , Cholera Toxin , Cinacalcet/metabolism , Colon/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Diarrhea/metabolism , Enterotoxins , Escherichia coli , Escherichia coli Proteins , Female , Humans , Hyperparathyroidism/drug therapy , Intestinal Mucosa/metabolism , Intestines/drug effects , Male , Mice
6.
Cell Physiol Biochem ; 53(2): 429-438, 2019.
Article in English | MEDLINE | ID: mdl-31424183

ABSTRACT

BACKGROUND/AIMS: Chronic kidney disease-mineral bone disorder is a major complication affecting the vast majority of chronic kidney disease patients. A hallmark of the disorder is an altered parathyroid gland biology resulting in secondary hyperparathyroidism. This condition is widely treated by calcimimetics like cinacalcet which act by allosteric activation of the calcium sensing receptor. METHODS: Here, we present a linear multi-compartment model based on physiological principles such as first-pass metabolism and protein binding, which captures all relevant pharmacokinetic parameters of cinacalcet. RESULTS: Due to the linear structure of the model, simulations are numerically stable and allow fast and accurate short or long-term predictions of cinacalcet concentrations in the body. CONCLUSION: The model compartments are physiological meaningful and can be easily adjusted to various conditions like impaired hepatic clearance or different drug administration regimens. Moreover, the model can be easily adapted to specific patient groups.


Subject(s)
Calcimimetic Agents/pharmacokinetics , Cinacalcet/pharmacokinetics , Models, Biological , Calcimimetic Agents/blood , Calcimimetic Agents/metabolism , Cinacalcet/blood , Cinacalcet/metabolism , Computer Simulation , Humans , Hyperparathyroidism, Secondary/blood , Hyperparathyroidism, Secondary/drug therapy , Hyperparathyroidism, Secondary/etiology , Hyperparathyroidism, Secondary/metabolism , Liver/metabolism , Protein Binding , Renal Insufficiency, Chronic/complications
7.
Article in English | MEDLINE | ID: mdl-27304782

ABSTRACT

In this investigation, a novel strategy for separation and quantitative determination of four metabolites of cinacalcet (M2a-Glu, M2b-Glu, M7-Gly, and M8-Gly) in human urine is suggested. The analytical assay is based on a pre-column derivation procedure of cinacalcet metabolites with 1-pyrenyldiazomethane (PDAM) as a fluorescent labeling reagent, and subsequently separation and quantitative determination with reverse-phase high-performance liquid chromatography (RP-HPLC) coupled with a fluorescence detector. Metabolites were separated on a Microsorb-MV 100-5 C18 chromatography column (250×4.6mm, 5µm) using acetate buffer (pH 3.5):methanol (30:70 v/v) as mobile phase at a flow rate of 1.0mLmin(-1). The method was fully validated in terms of linearity (r(2)>0.996; 1-10ngmL(-1)), precision (both intra-day and inter-day; RSD<6.2%), accuracy (92-110%), specificity, robustness (0.15%

Subject(s)
Calcimimetic Agents/urine , Chromatography, High Pressure Liquid/methods , Chromatography, Reverse-Phase/methods , Cinacalcet/urine , Calcimimetic Agents/analysis , Calcimimetic Agents/metabolism , Cinacalcet/analysis , Cinacalcet/metabolism , Fluorescence , Fluorescent Dyes/chemistry , Humans , Limit of Detection , Pyrenes/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...